1
|
Ng WK, Thanusha G, Chong PP, Chuah C. The Power of Antibodies: Advancing Biomarker-Based Disease Detection and Surveillance. Immunol Invest 2025:1-25. [PMID: 40256875 DOI: 10.1080/08820139.2025.2492246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
BACKGROUND Antibodies have long served as fundamental tools in disease diagnosis and surveillance. Their utility as biomarkers has expanded beyond infectious diseases to encompass a wide range of health conditions. OBJECTIVES This review aims to explore recent advancements in antibody biomarker discovery and their applications in diagnosing and monitoring diverse health conditions. It also examines the role of antibody surveillance in public health and epidemiological studies. METHODS A comprehensive analysis of recent literature was conducted, focusing on studies that identify and characterize disease-specific antibodies. Particular attention was given to their relevance in autoimmune diseases, infections, cancers, and neurological disorders. CONTENT The review highlights disease-specific antibody biomarkers and their clinical significance. It also discusses the utility and challenges of antibody-based surveillance in assessing disease prevalence, tracking immunity trends, and supporting One Health strategies. CONCLUSIONS Recent advancements in antibody biomarker discovery demonstrate significant potential in improving early diagnosis, personalized treatment, and population-level health management. Antibody surveillance continues to play a pivotal role in guiding public health responses and understanding disease dynamics.
Collapse
Affiliation(s)
- Woei Kean Ng
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| | - Gunasegran Thanusha
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Candy Chuah
- Unit of Microbiology, Faculty of Medicine, AIMST University, Bedong, Malaysia
| |
Collapse
|
2
|
Gentilotti E, Canziani LM, Caponcello MG, Azzini AM, Savoldi A, De Nardo P, Palacios-Baena ZR, Tazza B, Caroccia N, Marchetti G, Antinori A, Giannella M, Rodríguez-Baño J, Tacconelli E. ORCHESTRA Delphi consensus: diagnostic and therapeutic management of Post-COVID-19 condition in vulnerable populations. Clin Microbiol Infect 2025:S1198-743X(25)00172-7. [PMID: 40252805 DOI: 10.1016/j.cmi.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Post-COVID condition (PCC) remains poorly understood, especially in clinically vulnerable groups. Within the ORCHESTRA Project, we applied the Delphi approach to drive recommendations for the diagnosis, management, and prevention of PCC in people living with HIV (PWH) and patients affected by rheumatological diseases (RD) and haematological malignancies (HM). METHODS Based on literature review, three areas of interest in PCC in PWH, HM, and RD were identified: 1) features and risk factors; 2) diagnosis and management; and 3) prevention. A three-round Delphi anonymous survey consisting of 15 questions was conducted including 69 experts. Consensus was measured by the 6-point Likert scale categorised into four tiers: strong disagreement, moderate disagreement, moderate agreement, and strong agreement. Statements were generated on questions achieving consensus. RESULTS Eleven statements were generated: six on features and risk factors of PCC in clinically vulnerable populations, two on diagnosis and management, and three on prevention. Chronic fatigue was identified as the most frequent presentation of PCC in PWH and RD populations. A different case definition of PCC is required for RD population, as symptoms of PCC and autoimmune disorders may overlap. Risk factors for PCC include age>65, severity of COVID-19, and female sex; this latter is also associated with increased smell/taste impairment. A clinical assessment or a routine laboratory test performed three months after acute infection is not suggested to diagnose PCC in PWH. PWH and RD should be screened to exclude additional autoimmune disorders in case of chronic fatigue/arthralgia of new onset. Full-course vaccination and early treatment for COVID-19 should be promoted to prevent PCC, while corticosteroids during acute infection are not recommended. CONCLUSION Diagnosis, management and prevention of PCC are still under discussion. This Delphi offers valuable insights on PCC in selected clinically vulnerable populations and suggests a tailored approach in vulnerable populations.
Collapse
Affiliation(s)
- Elisa Gentilotti
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Lorenzo Maria Canziani
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.
| | - Maria Giulia Caponcello
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Hospital Universitario Virgen MacarenaDepartamento de Medicina, Universidad de SevillaCIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Maria Azzini
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Alessia Savoldi
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Pasquale De Nardo
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Zaira R Palacios-Baena
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Hospital Universitario Virgen MacarenaDepartamento de Medicina, Universidad de SevillaCIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Beatrice Tazza
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Natascia Caroccia
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giulia Marchetti
- . Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Andrea Antinori
- . Clinical Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Maddalena Giannella
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; . Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Jesús Rodríguez-Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Hospital Universitario Virgen MacarenaDepartamento de Medicina, Universidad de SevillaCIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
3
|
Nair CV, Krishnakumar M, Gutjahr G, Kulirankal KG, Moni M, Sathyapalan DT. Early biomarkers in hospitalized patients as predictors of post-acute sequelae of SARS-CoV-2 infection: a one-year cohort study. BMC Infect Dis 2025; 25:398. [PMID: 40128677 PMCID: PMC11931858 DOI: 10.1186/s12879-025-10619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/07/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Post-acute sequelae of SARS-CoV-2 infection (PASC) represent a significant challenge in patient care, with symptoms persisting beyond three month's post-recovery. This study aimed to evaluate the incidence of PASC at one year post-COVID-19 and identify predictive biomarkers and comorbidities for effective risk stratification. METHODS A cohort of 120 adult patients, including 50 intensive care and 70 non-intensive care patients, was followed up at two weeks, six weeks, and one-year post-discharge using structured questionnaires. The study integrated comorbidities and laboratory biomarkers to forecast the risk for PASC. RESULTS The median age of participants was 56 years, with 40% having moderate to severe comorbidities. A year post-recovery, 32.8% exhibited post COVID-19 conditions. The most common symptoms were constitutional (16%), respiratory (8.4%), and neuropsychiatric (2.5%). Bayesian network analysis indicated significant correlations between constitutional symptoms, rehospitalisation, and biomarkers including C-reactive protein, lactate-dehydrogenase, ferritin, and albumin. CONCLUSION This study highlights the prolonged impact of PASC, one-year post infection. It highlights the role of specific biomarkers such as C-reactive protein, lactate-dehydrogenase, ferritin, and albumin in tailoring individual patient care by advancing understanding in post-COVID-19 symptoms prediction. Our findings support the need for further research to refine these insights, which are pivotal for the ongoing care of patients in the aftermath of COVID-19.
Collapse
Affiliation(s)
- Chithira V Nair
- Division of Infectious Diseases, Department of General Medicine, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Malavika Krishnakumar
- Department of Health Sciences Research, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Georg Gutjahr
- Department of Health Sciences Research, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Kiran G Kulirankal
- Division of Infectious Diseases, Department of General Medicine, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Merlin Moni
- Division of Infectious Diseases, Department of General Medicine, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Dipu T Sathyapalan
- Division of Infectious Diseases, Department of General Medicine, Amrita Institute of Medical Science and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| |
Collapse
|
4
|
Masuda Y, Otsuka Y, Tokumasu K, Honda H, Sakurada Y, Matsuda Y, Nakano Y, Takase R, Omura D, Hasegawa T, Ueda K, Otsuka F. Interrelationships Between Plasma Levels of Brain Natriuretic Peptide and Prolonged Symptoms Due to Long COVID. J Clin Med 2025; 14:817. [PMID: 39941487 PMCID: PMC11818701 DOI: 10.3390/jcm14030817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Objectives: Evidence for the usefulness of biomarkers that aid in diagnosis, assessment of severity, and prediction of prognosis in patients with long COVID is limited. The aim of this study was to clarify the characteristics of brain natriuretic peptide (BNP) in long COVID. Methods: We conducted a retrospective observational study of patients who visited the COVID-19 aftercare outpatient clinic at Okayama University Hospital from February 2021 to April 2024. Results: A total of 428 patients were enrolled in this study, and the patients were divided into a group with normal BNP (n = 314, ≤18.4 pg/mL) and a group with increased BNP (n = 114, >18.4 pg/mL). The long COVID group with increased BNP had a higher proportion of females (44.3% vs. 73.7%, p < 0.01) and an older median age (38 vs. 51 years, p < 0.01). Fatigue and brain fog were commonly manifested in both groups, while dyspnea was a more frequent complaint in the group with increased BNP. Various symptoms including fatigue, palpitations, and taste and/or olfactory disorders were associated with elevated BNP (23 to 24 pg/mL). Memory impairment was also linked to higher BNP (OR: 2.36, p = 0.05). In long COVID patients, plasma BNP elevation appears to be more pronounced in females and is often related to cardiogenic factors, in which inflammatory responses are also involved. Conclusions: Plasma BNP measurement may be useful for evaluating the severity of long COVID, especially in female patients and those with respiratory symptoms and/or memory impairment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (Y.M.); (Y.O.); (K.T.); (H.H.); (Y.S.); (Y.M.); (Y.N.); (R.T.); (D.O.); (T.H.); (K.U.)
| |
Collapse
|
5
|
Iqbal NT, Khan H, Khalid A, Mahmood SF, Nasir N, Khanum I, de Siqueira I, Van Voorhis W. Chronic inflammation in post-acute sequelae of COVID-19 modulates gut microbiome: a review of literature on COVID-19 sequelae and gut dysbiosis. Mol Med 2025; 31:22. [PMID: 39849406 PMCID: PMC11756069 DOI: 10.1186/s10020-024-00986-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/01/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Long COVID or Post-acute sequelae of COVID-19 is an emerging syndrome, recognized in COVID-19 patients who suffer from mild to severe illness and do not recover completely. Most studies define Long COVID, through symptoms like fatigue, brain fog, joint pain, and headache prevailing four or more weeks post-initial infection. Global variations in Long COVID presentation and symptoms make it challenging to standardize features of Long COVID. Long COVID appears to be accompanied by an auto-immune multi-faceted syndrome where the virus or viral antigen persistence causes continuous stimulation of the immune response, resulting in multi-organ immune dysregulation. MAIN TEXT This review is focused on understanding the risk factors of Long COVID with a special emphasis on the dysregulation of the gut-brain axis. Two proposed mechanisms are discussed here. The first mechanism is related to the dysfunction of angiotensin-converting enzyme 2 receptor due to Severe Acute Respiratory Syndrome Corona Virus 2 infection, leading to impaired mTOR pathway activation, reduced AMP secretion, and causing dysbiotic changes in the gut. Secondly, gut-brain axis dysregulation accompanied by decreased production of short-chain fatty acids, impaired enteroendocrine cell function, and increased leakiness of the gut, which favors translocation of pathogens or lipopolysaccharide in circulation causing the release of pro-inflammatory cytokines. The altered Hypothalamic-Pituitary-Adrenal axis is accompanied by the reduced level of neurotransmitter, and decreased stimulation of the vagus nerve, which may cause neuroinflammation and dysregulation of serum cortisol levels. The dysbiotic microbiome in Long COVID patients is characterized by a decrease in beneficial short chain fatty acid-producing bacteria (Faecalibacterium, Ruminococcus, Dorea, and Bifidobacterium) and an increase in opportunistic bacteria (Corynebacterium, Streptococcus, Enterococcus). This dysbiosis is transient and may be impacted by interventions including probiotics, and dietary supplements. CONCLUSIONS Further studies are required to understand the geographic variation, racial and ethnic differences in phenotypes of Long COVID, the influence of viral strains on existing and emerging phenotypes, to explore long-term effects of gut dysbiosis, and gut-brain axis dysregulation, as well as the potential role of diet and probiotics in alleviating those symptoms.
Collapse
Affiliation(s)
- Najeeha Talat Iqbal
- Department of Biological and Biomedical Sciences, Department of Pediatrics and Child Health, Aga Khan University, Stadium Road, P. O Box 3500, Karachi, 74800, Pakistan.
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan.
| | - Hana Khan
- Undergraduate Medical Education (UGME), Year II, Aga Khan University, Karachi, Pakistan
| | - Aqsa Khalid
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Nosheen Nasir
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | | | - Wes Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, USA
| |
Collapse
|
6
|
Gonzalez-Ochoa AJ, Szolnoky G, Hernandez-Ibarra AG, Fareed J. Treatment with Sulodexide Downregulates Biomarkers for Endothelial Dysfunction in Convalescent COVID-19 Patients. Clin Appl Thromb Hemost 2025; 31:10760296241297647. [PMID: 39763448 PMCID: PMC11705351 DOI: 10.1177/10760296241297647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Persistent elevation of biomarkers associated with endothelial dysfunction in convalescent COVID-19 patients has been linked to an increased risk of long-term cardiovascular complications, including long COVID syndrome. Sulodexide, known for its vascular endothelial affinity, has demonstrated pleiotropic protective properties. This study aims to evaluate the impact of sulodexide on serum levels of endothelial dysfunction biomarkers in patients during the convalescent phase of COVID-19. METHODS We conducted a double-blind, single-center, randomized, placebo-controlled trial in Mexico, comparing sulodexide (250 LRU orally, twice daily) with placebo over 8 weeks in adult patients during early COVID-19 convalescence. Differences in serum biomarkers between the groups were analyzed using repeated measures and post hoc tests, with Thrombomodulin (TM) as the primary endpoint. RESULTS Among 206 analyzed patients (103 in each group), at week 8, the sulodexide group exhibited significantly lower mean levels of Thrombomodulin (TM) (25.2 ± 7.9 ng/mL vs 29.9 ± 14.7 ng/mL, P = .03), von Willebrand Factor (vWF) (232 ± 131 U/dL vs 266 ± 122 U/dL, P = .02) and Interleukin-6 (IL-6) (12.5 ± 13.2 pg/mL vs 16.2 ± 16.5 pg/mL, P = .03) compared to the placebo group. D-dimer and C reactive protein (CRP) in the sulodexide group were also lowered. No significant differences were observed for P-selectin, fibrinogen, VCAM-1, or ICAM-1 levels. CONCLUSIONS Patients in the convalescent phase of COVID-19 who received sulodexide for eight weeks showed a reduction in TM, vWF, D-dimer, CRP, and IL-6 serum levels compared to placebo. These findings suggest a potential protective effect of sulodexide against thromboinflammation and endothelial damage.
Collapse
Affiliation(s)
- Alejandro J Gonzalez-Ochoa
- Vascular Surgery Department, Centro Médico del Noroeste, San Luis Rio Colorado, Sonora, México
- Vascular and Endovascular Surgery department, CLINEDEM, San Luis Rio Colorado, Sonora, México
| | - Gyozo Szolnoky
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | | | - Jawed Fareed
- Hemostasis and Thrombosis Research Laboratories, Loyola University Medical Center, Maywood, Illinois, USA
| |
Collapse
|
7
|
Huang HP, Wang HW, Cheng CH, Chang YS, Yeh TK, Huang WH, Liu CW, Chang CH, Liu PY, Yen YC, Tseng CH. Post-COVID-19 Condition and Pulmonary Embolism. J Multidiscip Healthc 2024; 17:6153-6159. [PMID: 39741525 PMCID: PMC11687424 DOI: 10.2147/jmdh.s492159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/09/2024] [Indexed: 01/03/2025] Open
Abstract
Purpose Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes post-acute sequelae of coronavirus disease 2019 (COVID-19), including pulmonary vasculopathy, increasing thrombotic risk. Screening and treating survivors are essential to reduce associated disabilities. We aim to investigate the clinical characteristics of patients with post-COVID-19 condition and pulmonary embolism, as well as their health-related quality of life one year after COVID-19 diagnosis. Patients and Methods In our study, we analyzed nine cases of post-COVID-19 condition and pulmonary embolism in a tertiary hospital in Taiwan. Patient characteristics, including age, sex, symptoms, and outcomes, were recorded. One year post-diagnosis, patients underwent follow-up with lab tests, chest X-rays, electrocardiograms, and health-related quality of life (HRQOL) assessments using the EuroQoL 5-Dimension 5-Level (EQ-5D-5L) tool. Results Post-COVID-19 condition with pulmonary embolism predominantly affects females. Common symptoms include breathlessness and chest pain, and a lung perfusion scan may be useful for diagnosis. The one-year follow-up of five patients mainly showed normal lab results and no active lung lesions. The mean EQ-5D score was 0.928 ± 0.119, and the EQ visual analogue scale (EQ-VAS) was 85.00 ± 11.18. Pain/discomfort was the most reported issue. Conclusion This research reveals that post-COVID-19 condition with pulmonary embolism affects a significant number of women and those with preexisting conditions like cancer or diabetes. Symptoms overlap also complicates diagnosis. Although many lab results were normal, the elevated risk of pulmonary embolism persists. Lower utility scores and increased anxiety highlight the need for targeted interventions and mental health support. Our study underscores the importance of monitoring pulmonary embolism in patients after COVID-19 infection, given the significant impact on post-COVID-19 condition and overall health outcomes.
Collapse
Affiliation(s)
- Hsien-Po Huang
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Genomic Center for Infectious Diseases, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsiu-Wen Wang
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Hsin Cheng
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Shan Chang
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ting-Kuang Yeh
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Genomic Center for Infectious Diseases, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wei-Hsuan Huang
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Wei Liu
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Hung Chang
- Department of Computer Science and Communication Engineering, Providence University, Taichung, Taiwan
| | - Po-Yu Liu
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Genomic Center for Infectious Diseases, Taichung Veterans General Hospital, Taichung, Taiwan
- Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Chun Yen
- Division of Clinical Informatics, Department of Digital Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chien-Hao Tseng
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
8
|
Meirman TD, Shapira B, Balicer RD, Rokach L, Dagan N. Trends of common laboratory biomarkers after SARS-CoV-2 infection. J Infect 2024; 89:106318. [PMID: 39423876 DOI: 10.1016/j.jinf.2024.106318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Most studies that explore the long-term effects of COVID-19 are based on subjectively reported symptoms, while laboratory-measured biomarkers are mainly examined in studies of relatively small cohorts. This study investigates the long-term effects of SARS-CoV-2 infection on common laboratory biomarkers. METHODS We utilized a retrospective cohort of SARS-CoV-2 infected individuals and rigorously matched controls based on demographic and clinical characteristics, examining 63 common laboratory biomarkers. Additional lab-specific cohorts were matched with an additional criterion of baseline biomarker values. Differences in biomarkers over a 12-month follow-up were analyzed using standardized mean difference-in-differences. RESULTS The general cohort included 361,061 matched pairs, with 26M laboratory results. The effects on most biomarkers lasted 1-4 months and were consistent with anticipated changes after acute viral infections. Some biomarkers presented prolonged effects, consistent across the general and lab-specific cohorts. One group of such findings included a 7-8 month decrease in WBC counts, mainly driven by decreased counts of neutrophils, monocytes, and basophils. Potassium levels were decreased for 3-5 months. Vaccinated individuals' data suggested potentially smaller effects on WBCs, but cohort sizes limited this analysis. CONCLUSIONS This study explores SARS-CoV-2 infection effects on common laboratory biomarkers, characterizing the direction and duration of these effects on the largest infected cohort to date. The effects of most biomarkers resolve in the first months following infection. The most notable longer-lasting effects involved the immune system. Further research is required to characterize the magnitude of these effects among specific individuals.
Collapse
Affiliation(s)
| | | | - Ran D Balicer
- Ben-Gurion University of the Negev, Israel; Clalit Research Institute, Israel; The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute.
| | | | - Noa Dagan
- Ben-Gurion University of the Negev, Israel; Clalit Research Institute, Israel; The Ivan and Francesca Berkowitz Family Living Laboratory Collaboration at Harvard Medical School and Clalit Research Institute.
| |
Collapse
|
9
|
Korth J, Steenblock C, Walther R, Barbir M, Husung M, Velthof A. A Single-Center Pilot Study of Therapeutic Apheresis in Patients with Severe Post-COVID Syndrome. Horm Metab Res 2024; 56:869-874. [PMID: 39653042 DOI: 10.1055/a-2445-8593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
After the COVID-19 pandemic, many patients have reported chronic fatigue and severe post-exertional malaise, with symptoms similar to those of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). The accumulation of agonistic receptor autoantibodies targeting beta-adrenergic (β1 and β2) and muscarinic (M3 and M4) neurotransmitter receptors may play a crucial role in the pathomechanism of both ME/CFS and post-COVID conditions. Therapeutic apheresis has been suggested as an effective treatment option for alleviating and mitigating symptoms in this desperate group of patients. In this single-center pilot study, we analyzed autoantibodies in a cohort of 20 post-COVID patients before and after therapeutic apheresis. Apheresis resulted in a decline of β1 or β2 adrenergic receptor antibodies in all patients. Additionally, the majority of patients experienced a concurrent reduction in symptoms such as fatigue, physical activity restrictions, myalgia, post-exertional malaise, and concentration disorders. This study clearly demonstrates an association between autoantibodies and the clinical improvement of post-COVID patients. Even if future sham-controlled trials do not show a positive outcome, extracorporeal apheresis may still be valuable for this patient group by temporarily improving microperfusion and symptoms. Success in restoring patients to work and normal life, as observed in many individuals after therapeutic apheresis, should be recognized. Therefore, we believe that extracorporeal therapeutic apheresis, as part of a multimodal treatment, should be considered an early intervention for postinfectious syndromes in selected patients.
Collapse
Affiliation(s)
| | - Charlotte Steenblock
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Romy Walther
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mahmoud Barbir
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Cardiology, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust Hospital, Harefield, United Kingdom of Great Britain and Northern Ireland
| | | | | |
Collapse
|
10
|
Oropeza-Valdez JJ, Padron-Manrique C, Vázquez-Jiménez A, Soberon X, Resendis-Antonio O. Exploring metabolic anomalies in COVID-19 and post-COVID-19: a machine learning approach with explainable artificial intelligence. Front Mol Biosci 2024; 11:1429281. [PMID: 39314212 PMCID: PMC11417410 DOI: 10.3389/fmolb.2024.1429281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has led to significant challenges worldwide, including diverse clinical outcomes and prolonged post-recovery symptoms known as Long COVID or Post-COVID-19 syndrome. Emerging evidence suggests a crucial role of metabolic reprogramming in the infection's long-term consequences. This study employs a novel approach utilizing machine learning (ML) and explainable artificial intelligence (XAI) to analyze metabolic alterations in COVID-19 and Post-COVID-19 patients. Samples were taken from a cohort of 142 COVID-19, 48 Post-COVID-19, and 38 control patients, comprising 111 identified metabolites. Traditional analysis methods, like PCA and PLS-DA, were compared with ML techniques, particularly eXtreme Gradient Boosting (XGBoost) enhanced by SHAP (SHapley Additive exPlanations) values for explainability. XGBoost, combined with SHAP, outperformed traditional methods, demonstrating superior predictive performance and providing new insights into the metabolic basis of the disease's progression and aftermath. The analysis revealed metabolomic subgroups within the COVID-19 and Post-COVID-19 conditions, suggesting heterogeneous metabolic responses to the infection and its long-term impacts. Key metabolic signatures in Post-COVID-19 include taurine, glutamine, alpha-Ketoglutaric acid, and LysoPC a C16:0. This study highlights the potential of integrating ML and XAI for a fine-grained description in metabolomics research, offering a more detailed understanding of metabolic anomalies in COVID-19 and Post-COVID-19 conditions.
Collapse
Affiliation(s)
- Juan José Oropeza-Valdez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Cristian Padron-Manrique
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Xavier Soberon
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Colonia Chamilpa, Cuernavaca, México
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- Coordinación de la Investigación Científica – Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
11
|
Erlandson KM, Geng LN, Selvaggi CA, Thaweethai T, Chen P, Erdmann NB, Goldman JD, Henrich TJ, Hornig M, Karlson EW, Katz SD, Kim C, Cribbs SK, Laiyemo AO, Letts R, Lin JY, Marathe J, Parthasarathy S, Patterson TF, Taylor BD, Duffy ER, Haack M, Julg B, Maranga G, Hernandez C, Singer N, Han J, Pemu P, Brim H, Ashktorab H, Charney AW, Wisnivesky J, Lin J, Chu HY, Go M, Singh U, Levitan EB, Goepfert PA, Nikolich JŽ, Hsu H, Peluso MJ, Kelly JD, Okumura M, Flaherman VJ, Quigley JG, Krishnan JA, Scholand MB, Hess R, Metz TD, Costantine MM, Rouse DJ, Taylor BS, Goldberg MP, Marshall GD, Wood J, Warren D, Horwitz L, Foulkes AS, McComsey GA. Differentiation of Prior SARS-CoV-2 Infection and Postacute Sequelae by Standard Clinical Laboratory Measurements in the RECOVER Cohort. Ann Intern Med 2024; 177:1209-1221. [PMID: 39133923 PMCID: PMC11408082 DOI: 10.7326/m24-0737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND There are currently no validated clinical biomarkers of postacute sequelae of SARS-CoV-2 infection (PASC). OBJECTIVE To investigate clinical laboratory markers of SARS-CoV-2 and PASC. DESIGN Propensity score-weighted linear regression models were fitted to evaluate differences in mean laboratory measures by prior infection and PASC index (≥12 vs. 0). (ClinicalTrials.gov: NCT05172024). SETTING 83 enrolling sites. PARTICIPANTS RECOVER-Adult cohort participants with or without SARS-CoV-2 infection with a study visit and laboratory measures 6 months after the index date (or at enrollment if >6 months after the index date). Participants were excluded if the 6-month visit occurred within 30 days of reinfection. MEASUREMENTS Participants completed questionnaires and standard clinical laboratory tests. RESULTS Among 10 094 participants, 8746 had prior SARS-CoV-2 infection, 1348 were uninfected, 1880 had a PASC index of 12 or higher, and 3351 had a PASC index of zero. After propensity score adjustment, participants with prior infection had a lower mean platelet count (265.9 × 109 cells/L [95% CI, 264.5 to 267.4 × 109 cells/L]) than participants without known prior infection (275.2 × 109 cells/L [CI, 268.5 to 282.0 × 109 cells/L]), as well as higher mean hemoglobin A1c (HbA1c) level (5.58% [CI, 5.56% to 5.60%] vs. 5.46% [CI, 5.40% to 5.51%]) and urinary albumin-creatinine ratio (81.9 mg/g [CI, 67.5 to 96.2 mg/g] vs. 43.0 mg/g [CI, 25.4 to 60.6 mg/g]), although differences were of modest clinical significance. The difference in HbA1c levels was attenuated after participants with preexisting diabetes were excluded. Among participants with prior infection, no meaningful differences in mean laboratory values were found between those with a PASC index of 12 or higher and those with a PASC index of zero. LIMITATION Whether differences in laboratory markers represent consequences of or risk factors for SARS-CoV-2 infection could not be determined. CONCLUSION Overall, no evidence was found that any of the 25 routine clinical laboratory values assessed in this study could serve as a clinically useful biomarker of PASC. PRIMARY FUNDING SOURCE National Institutes of Health.
Collapse
Affiliation(s)
- Kristine M. Erlandson
- University of Colorado, Anschutz Medical Campus; Department of Medicine, Division of Infectious Diseases; Aurora, CO
| | - Linda N. Geng
- Stanford University; Department of Medicine; Stanford, CA
| | | | | | - Peter Chen
- Cedars-Sinai Medical Center; Department of Medicine. Division of Pulmonary and Critical Care Medicine; New York, NY
- Women’s Guild Lung Institute at Cedars-Sinai Medical Center; New York, NY
| | - Nathan B. Erdmann
- University of Alabama at Birmingham, Department of Medicine, Division of Infectious Diseases, Birmingham, AL
| | - Jason D. Goldman
- Swedish Center for Research and Innovation, Providence Swedish Medical Center; Seattle, WA
- University of Washington, Division of Allergy and Infectious Diseases; Seattle, WA
| | - Timothy J. Henrich
- University of California San Francisco, Division of Experimental Medicine, San Francisco, CA
| | - Mady Hornig
- CORe Community Inc., New York, NY
- Columbia University Mailman School of Public Health, Department of Epidemiology, New York, NY
| | | | - Stuart D. Katz
- NYU Grossman School of Medicine, Department of Medicine, New York, NY
| | - C. Kim
- RECOVER Initiative, New York, NY
| | - Sushma K. Cribbs
- Emory University, School of Medicine, Department of Medicine, Atlanta, GA
- Atlanta Veterans Affairs Medical Center; Atlanta, Georgia
| | - Adeyinka O. Laiyemo
- Howard University College of Medicine, Department of Medicine, Division of Gastroenterology, Washington DC
| | | | - Janet Y. Lin
- University of Illinois Chicago, Department of Emergency Medicine, Chicago, IL
| | - Jai Marathe
- Boston University Medical Campus, Department of Medicine, Division of Infectious Diseases, Boston, MA
| | | | - Thomas F. Patterson
- University of Texas Health San Antonio, Department of Medicine, San Antonio, Texas
| | - Brittany D. Taylor
- RECOVER Initiative, New York, NY
- American Heart Association, Health Strategies, Atlanta, GA
| | | | - Monika Haack
- Beth Israel Deaconess Medical Center, Department of Neurology; Boston, MA
| | - Boris Julg
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard; Boston, MA
| | - Gabrielle Maranga
- NYU Grossman School of Medicine, Department of Population Health, New York, NY
| | - Carla Hernandez
- Case Western Reserve University, Departments of Pediatrics and Medicine, Cleveland, OH
| | - Nora Singer
- Case Western Reserve University, Departments of Pediatrics and Medicine, Cleveland, OH
- Case Western Reserve University, Division of Rheumatology, Cleveland, OH
| | - Jenny Han
- Emory University, School of Medicine, Department of Medicine, Atlanta, GA
- Grady Hospital, Atlanta, GA
| | - Priscilla Pemu
- Morehouse School of Medicine, Department of Medicine, Atlanta, GA
| | - Hassan Brim
- Howard University, Department of Pathology, Washington, DC
| | | | | | - Juan Wisnivesky
- Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - Jenny Lin
- Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - Helen Y. Chu
- University of Washington, Division of Global Health, Seattle, WA
| | - Minjoung Go
- Stanford University; Department of Medicine; Stanford, CA
| | - Upinder Singh
- Stanford University; Department of Medicine; Stanford, CA
| | - Emily B. Levitan
- University of Alabama at Birmingham, Department of Epidemiology, Birmingham, AL
| | - Paul A. Goepfert
- University of Alabama at Birmingham, Department of Medicine, Division of Infectious Diseases, Birmingham, AL
| | - Janko Ž. Nikolich
- University of Arizona College of Medicine-Tucson, Department of Immunobiology, Tucson, AZ
- Arizona Center on Aging, Tucson, AZ
| | - Harvey Hsu
- Banner University Medical Center, Tucson, AZ
| | - Michael J. Peluso
- University of California San Francisco, Department of Medicine, Division of Infectious Diseases, San Francisco, CA
| | - J. Daniel Kelly
- University of California San Francisco, Department of Medicine, Division of Infectious Diseases, San Francisco, CA
| | - Megumi Okumura
- University of California San Francisco, Departments of Medicine and Pediatrics, San Francisco, CA
| | - Valerie J Flaherman
- University of California San Francisco, Department of Pediatrics, San Francisco, CA
| | - John G. Quigley
- University of Illinois Chicago, Department of Medicine, Division of Hematology/Oncology, Chicago, IL
| | | | - Mary Beth Scholand
- Spencer Fox Eccles School of Medicine at the University of Utah, Department of Medicine, Salt Lake City, UT
| | - Rachel Hess
- Spencer Fox Eccles School of Medicine at the University of Utah, Department of Medicine, Salt Lake City, UT
| | - Torri D. Metz
- University of Utah, Department of Obstetrics and Gynecology, Salt Lake City, UT
| | - Maged M. Costantine
- The Ohio State University, Division of Maternal Fetal Medicine, Columbus, OH
| | - Dwight J Rouse
- Brown University, Department of Obstetrics and Gynecology, Providence, RI
| | - Barbara S. Taylor
- University of Texas Health San Antonio, Department of Medicine, San Antonio, Texas
| | - Mark P. Goldberg
- University of Texas Health San Antonio, Department of Neurology, San Antonio, Texas
| | - Gailen D. Marshall
- University of Mississippi Medical Center, Department of Medicine, Jackson, MS
| | - Jeremy Wood
- The Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - David Warren
- University of Nebraska Medical Center, Department of Neurological Sciences, Omaha, NE
| | - Leora Horwitz
- NYU Grossman School of Medicine, Department of Population Health, New York, NY
- Center for Healthcare Innovation and Delivery Science, NYU Langone Health, New York, NY
| | | | - Grace A McComsey
- Case Western Reserve University, Departments of Pediatrics and Medicine, Cleveland, OH
| |
Collapse
|
12
|
Galúcio VCA, de Menezes DC, Chaves ECR, van den Berg AVS, de Lima PDL, da Costa Vasconcelos PF, Quaresma JAS, Falcão LFM. Laboratory profiling of patients with long COVID in the Brazilian Amazon region: A cross-sectional study. J Med Virol 2024; 96:e29828. [PMID: 39081145 DOI: 10.1002/jmv.29828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/16/2024] [Indexed: 01/25/2025]
Abstract
The condition commonly referred to as long coronavirus disease (COVID) is characterized by the continuation of symptoms, sometimes accompanied by new symptoms that persist after the resolution of acute coronavirus disease 2019 (COVID-19). This observational cross-sectional study investigated 332 patients with long COVID in the Brazilian Amazon region. The study aimed to elucidate the systemic interactions associated with long COVID by compiling the findings related to hematological, coagulation, immunological, metabolic, hepatic, renal, and muscular profiles. Participants with long COVID were identified using rigorous criteria and underwent thorough laboratory examinations. The obtained data were subsequently analyzed, allowing for comparisons, associations, and correlations between findings within distinct groups in the study. Significant associations were observed between hospitalization during the acute phase and persistent laboratory abnormalities, suggesting a potential link between acute severity and long-term effects. Notably, individuals with long COVID for over a year exhibited elevated levels of monocytes, prolonged prothrombin times, reduced prothrombin activity, high levels of lactate dehydrogenase, and an increased frequency of qualitative C-reactive protein detection. This study provides valuable insights into the laboratory risk profile of patients with long COVID, particularly in the unique context of the Amazon region, where patients exhibit persistent symptoms lasting up to 1261 days.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Juarez Antônio Simões Quaresma
- Centre for Biological Health Sciences, State University of Pará (UEPA), Belém, Brazil
- School of Medicine, São Paulo University (USP), São Paulo, Brazil
- Tropical Medicine Centre, Federal University of Pará (UFPA), Belém, Brazil
| | - Luiz Fábio Magno Falcão
- Centre for Biological Health Sciences, State University of Pará (UEPA), Belém, Brazil
- School of Medicine, São Paulo University (USP), São Paulo, Brazil
| |
Collapse
|
13
|
Abo Elyazed TI, Abd El-Hakim AAEM, Saleh OI, Sonbol MMF, Eid HA, Moazen E, Alhassoon MH, Elfeky SEF. Diaphragmatic strengthening exercises for patients with post COVID-19 condition after mild-to-moderate acute COVID-19 infection: a randomized controlled study. J Rehabil Med 2024; 56:jrm25491. [PMID: 38860716 PMCID: PMC11182030 DOI: 10.2340/jrm.v56.25491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVE To assess the clinical effects of incentive spirometry (IS) and diaphragmatic breathing (DB) in patients with post COVID-19 condition and diaphragmatic dysfunction as compared with the standard care alone. METHODS The present longitudinal randomized study included 60 patients with post COVID-19 condition and diaphragmatic dysfunction. Patients were equally randomized to receive standard care plus IS (G1), standard care plus DB (G2) or standard care alone (G3) for 8 weeks. The primary outcome is clinical improvement as evaluated by the modified Medical Research Council (mMRC) dyspnoea scale. RESULTS Comparison between the studied groups revealed significant improvement in G1 and G2 in all parameters at the end of follow-up. However, no significant improvement was found in G3. At the end of follow-up, 15 patients (75.0%) in G1, 11 patients (55.0%) in G2, and 3 patients (15.0%) in G3 showed improvement on the mMRC dyspnoea scale. Multivariate logistic regression analysis identified mild acute COVID-19 infection (p = 0.009), use of IS (p < 0.001), and use of DB (p = 0.023) as significant predictors of improvement on the mMRC dyspnoea scale. CONCLUSIONS IS or DB training in addition to the standard care in post COVID-19 condition was associated with better clinical improvement as compared with the standard care alone.
Collapse
Affiliation(s)
- Tamer I Abo Elyazed
- Physical Therapy For Internal Medicine Department, Faculty of Physical Therapy, Beni-Suef University, Ben-Suef, Egypt
| | | | - Ola I Saleh
- Diagnostic Radiology Department, Al-Azhar University, Cairo, Egypt
| | | | - Hoda Assad Eid
- Chest Diseases Department, Al-Azhar University, Cairo, Egypt
| | - Eman Moazen
- Chest Diseases Department, Al-Azhar University, Cairo, Egypt.
| | | | | |
Collapse
|
14
|
Rathod N, Kumar S, Chavhan R, Acharya S, Rathod S. Navigating the Long Haul: A Comprehensive Review of Long-COVID Sequelae, Patient Impact, Pathogenesis, and Management. Cureus 2024; 16:e60176. [PMID: 38868283 PMCID: PMC11167581 DOI: 10.7759/cureus.60176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/12/2024] [Indexed: 06/14/2024] Open
Abstract
Long COVID, characterized by persistent symptoms following a SARS-CoV-2 infection, presents a significant public health challenge with wide-ranging implications. This comprehensive review explores the epidemiology, clinical manifestations, pathogenesis, risk factors, diagnosis, patient impact, management strategies, and long-term prognosis of COVID. Despite a varied symptomatology that spans multiple organ systems, including respiratory, neurological, and cardiovascular systems, this condition is primarily associated with chronic inflammation and potential viral persistence. Prevalence varies, influenced by the initial infection severity, demographic factors, and pre-existing conditions. The review emphasizes the necessity for healthcare systems to adapt to the needs of long-COVID patients by developing standardized diagnostic criteria and personalized, multidisciplinary treatment approaches. Current research gaps and future directions are identified, highlighting the urgent need for further studies on pathophysiological mechanisms and effective therapeutic interventions. This review aims to inform healthcare providers, researchers, and policymakers, enhancing patient care and guiding ongoing and future research initiatives. The continuing global focus and collaborative efforts offer hope for improved outcomes for those affected by long COVID, marking an essential step towards addressing this emergent condition comprehensively.
Collapse
Affiliation(s)
- Nishant Rathod
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roma Chavhan
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sourya Acharya
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar Rathod
- Neurosurgery, Trivandrum Medical College, Thiruvananthapuram, IND
| |
Collapse
|
15
|
Domingo JC, Battistini F, Cordobilla B, Zaragozá MC, Sanmartin-Sentañes R, Alegre-Martin J, Cambras T, Castro-Marrero J. Association of circulating biomarkers with illness severity measures differentiates myalgic encephalomyelitis/chronic fatigue syndrome and post-COVID-19 condition: a prospective pilot cohort study. J Transl Med 2024; 22:343. [PMID: 38600563 PMCID: PMC11005215 DOI: 10.1186/s12967-024-05148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/30/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Accumulating evidence suggests that autonomic dysfunction and persistent systemic inflammation are common clinical features in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and long COVID. However, there is limited knowledge regarding their potential association with circulating biomarkers and illness severity in these conditions. METHODS This single-site, prospective, cross-sectional, pilot cohort study aimed to distinguish between the two patient populations by using self-reported outcome measures and circulating biomarkers of endothelial function and systemic inflammation status. Thirty-one individuals with ME/CFS, 23 individuals with long COVID, and 31 matched sedentary healthy controls were included. All study participants underwent non-invasive cardiovascular hemodynamic challenge testing (10 min NASA lean test) for assessment of orthostatic intolerance. Regression analysis was used to examine associations between outcome measures and circulating biomarkers in the study participants. Classification across groups was based on principal component and discriminant analyses. RESULTS Four ME/CFS patients (13%), 1 with long COVID (4%), and 1 healthy control (3%) presented postural orthostatic tachycardia syndrome (POTS) using the 10-min NASA lean test. Compared with matched healthy controls, ME/CFS and long COVID subjects showed higher levels of ET-1 (p < 0.05) and VCAM-1 (p < 0.001), and lower levels of nitrites (NOx assessed as NO2- + NO3-) (p < 0.01). ME/CFS patients also showed higher levels of serpin E1 (PAI-1) and E-selectin than did both long COVID and matched control subjects (p < 0.01 in all cases). Long COVID patients had lower TSP-1 levels than did ME/CFS patients and matched sedentary healthy controls (p < 0.001). As for inflammation biomarkers, both long COVID and ME/CFS subjects had higher levels of TNF-α than did matched healthy controls (p < 0.01 in both comparisons). Compared with controls, ME/CFS patients had higher levels of IL-1β (p < 0.001), IL-4 (p < 0.001), IL-6 (p < 0.01), IL-10 (p < 0.001), IP-10 (p < 0.05), and leptin (p < 0.001). Principal component analysis supported differentiation between groups based on self-reported outcome measures and biomarkers of endothelial function and inflammatory status in the study population. CONCLUSIONS Our findings revealed that combining biomarkers of endothelial dysfunction and inflammation with outcome measures differentiate ME/CFS and Long COVID using robust discriminant analysis of principal components. Further research is needed to provide a more comprehensive characterization of these underlying pathomechanisms, which could be promising targets for therapeutic and preventive strategies in these conditions.
Collapse
Affiliation(s)
- Joan Carles Domingo
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | - Federica Battistini
- Molecular Modelling and Bioinformatics Group, Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Begoña Cordobilla
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | | | - Ramón Sanmartin-Sentañes
- Division of Rheumatology, Clinical Unit in ME/CFS and Long COVID, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Jose Alegre-Martin
- Division of Rheumatology, Clinical Unit in ME/CFS and Long COVID, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain
| | - Trinitat Cambras
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, 08028, Spain.
| | - Jesus Castro-Marrero
- Division of Rheumatology, Research Unit in ME/CFS and Long COVID, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, 08035, Spain.
| |
Collapse
|
16
|
Goldstein DS. Post-COVID dysautonomias: what we know and (mainly) what we don't know. Nat Rev Neurol 2024; 20:99-113. [PMID: 38212633 DOI: 10.1038/s41582-023-00917-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Following on from the COVID-19 pandemic is another worldwide public health challenge that is referred to variously as long COVID, post-COVID syndrome or post-acute sequelae of SARS-CoV-2 infection (PASC). PASC comes in many forms and affects all body organs. This heterogeneous presentation suggests involvement of the autonomic nervous system (ANS), which has numerous roles in the maintenance of homeostasis and coordination of responses to various stressors. Thus far, studies of ANS dysregulation in people with PASC have been largely observational and descriptive, based on symptom inventories or objective but indirect measures of cardiovascular function, and have paid little attention to the adrenomedullary, hormonal and enteric nervous components of the ANS. Such investigations do not consider the syndromic nature of autonomic dysfunction. This Review provides an update on the literature relating to ANS abnormalities in people with post-COVID syndrome and presents a theoretical perspective on how the ANS might participate in common features of PASC.
Collapse
Affiliation(s)
- David S Goldstein
- Division of Intramural Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Agoston DV. Traumatic Brain Injury in the Long-COVID Era. Neurotrauma Rep 2024; 5:81-94. [PMID: 38463416 PMCID: PMC10923549 DOI: 10.1089/neur.2023.0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Major determinants of the biological background or reserve, such as age, biological sex, comorbidities (diabetes, hypertension, obesity, etc.), and medications (e.g., anticoagulants), are known to affect outcome after traumatic brain injury (TBI). With the unparalleled data richness of coronavirus disease 2019 (COVID-19; ∼375,000 and counting!) as well as the chronic form, long-COVID, also called post-acute sequelae SARS-CoV-2 infection (PASC), publications (∼30,000 and counting) covering virtually every aspect of the diseases, pathomechanisms, biomarkers, disease phases, symptomatology, etc., have provided a unique opportunity to better understand and appreciate the holistic nature of diseases, interconnectivity between organ systems, and importance of biological background in modifying disease trajectories and affecting outcomes. Such a holistic approach is badly needed to better understand TBI-induced conditions in their totality. Here, I briefly review what is known about long-COVID/PASC, its underlying-suspected-pathologies, the pathobiological changes induced by TBI, in other words, the TBI endophenotypes, discuss the intersection of long-COVID/PASC and TBI-induced pathobiologies, and how by considering some of the known factors affecting the person's biological background and the inclusion of mechanistic molecular biomarkers can help to improve the clinical management of TBI patients.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology, and Genetics, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Boyarchuk O, Volianska L. Autoimmunity and long COVID in children. Reumatologia 2024; 61:492-501. [PMID: 38322108 PMCID: PMC10839920 DOI: 10.5114/reum/176464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/06/2023] [Indexed: 02/08/2024] Open
Abstract
Numerous hypotheses regarding the pathogenetic mechanisms of long COVID have been proposed. Immune dysregulation and autoimmunity are among the leading hypotheses. In this article, we present two clinical cases of long COVID. The first case demonstrates the phenotype of long COVID with pain and musculoskeletal symptoms, which is often associated with autoimmunity and mimics systemic connective tissue diseases. In the second case, a high titer of antinuclear antibodies was observed after SARS-CoV-2 infection, but the clinical symptoms were limited to fever and headache. Only a comprehensive evaluation of clinical symptoms and thorough objective examination can confirm or exclude autoimmune diseases after a previous SARS-CoV-2 infection. A systematic search in the PubMed Medline database was carried out for studies focusing on immune dysregulation, autoimmunity, and its association with the clinical phenotype of long COVID. The question of the role of autoimmunity in the development of long COVID and the management approaches are discussed.
Collapse
Affiliation(s)
- Oksana Boyarchuk
- Department of Children's Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ukraine
| | - Liubov Volianska
- Department of Children's Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ukraine
| |
Collapse
|
19
|
Cezar R, Kundura L, André S, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. T4 apoptosis in the acute phase of SARS-CoV-2 infection predicts long COVID. Front Immunol 2024; 14:1335352. [PMID: 38235145 PMCID: PMC10791767 DOI: 10.3389/fimmu.2023.1335352] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Background As about 10% of patients with COVID-19 present sequelae, it is important to better understand the physiopathology of so-called long COVID. Method To this aim, we recruited 29 patients hospitalized for SARS-CoV-2 infection and, by Luminex®, quantified 19 soluble factors in their plasma and in the supernatant of their peripheral blood mononuclear cells, including inflammatory and anti-inflammatory cytokines and chemokines, Th1/Th2/Th17 cytokines, and endothelium activation markers. We also measured their T4, T8 and NK differentiation, activation, exhaustion and senescence, T cell apoptosis, and monocyte subpopulations by flow cytometry. We compared these markers between participants who developed long COVID or not one year later. Results None of these markers was predictive for sequelae, except programmed T4 cell death. T4 lymphocytes from participants who later presented long COVID were more apoptotic in culture than those of sequelae-free participants at Month 12 (36.9 ± 14.7 vs. 24.2 ± 9.0%, p = 0.016). Conclusions Our observation raises the hypothesis that T4 cell death during the acute phase of SARS-CoV-2 infection might pave the way for long COVID. Mechanistically, T4 lymphopenia might favor phenomena that could cause sequelae, including SARS-CoV-2 persistence, reactivation of other viruses, autoimmunity and immune dysregulation. In this scenario, inhibiting T cell apoptosis, for instance, by caspase inhibitors, could prevent long COVID.
Collapse
Affiliation(s)
- Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| | - Sonia André
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Ahn Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
- Laval University Research Center, Quebec City, QC, Canada
| | - Pierre Corbeau
- Immunology Department, Nîmes University Hospital, Nîmes, France
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| |
Collapse
|